7 research outputs found

    Post-CNS-inflammation expression of CXCL12 promotes the endogenous myelin/neuronal repair capacity following spontaneous recovery from multiple sclerosis-like disease

    Get PDF
    Background: Demyelination and axonal degeneration, hallmarks of multiple sclerosis (MS), are associated with the central nervous system (CNS) inflammation facilitated by C-X-C motif chemokine 12 (CXCL12) chemokine. Both in MS and in experimental autoimmune encephalomyelitis (EAE), the deleterious CNS inflammation has been associated with upregulation of CXCL12 expression in the CNS. We investigated the expression dynamics of CXCL12 in the CNS with progression of clinical EAE and following spontaneous recovery, with a focus on CXCL12 expression in the hippocampal neurogenic dentate gyrus (DG) and in the corpus callosum (CC) of spontaneously recovered mice, and its potential role in promoting the endogenous myelin/neuronal repair capacity. Methods: CNS tissue sections from mice with different clinical EAE phases or following spontaneous recovery and in vitro differentiated adult neural stem cell cultures were analyzed by immunofluorescent staining and confocal imaging for detecting and enumerating neuronal progenitor cells (NPCs) and oligodendrocyte precursor cells (OPCs) and for expression of CXCL12. Results: Our expression dynamics analysis of CXCL12 in the CNS with EAE progression revealed elevated CXCL12 expression in the DG and CC, which persistently increases following spontaneous recovery even though CNS inflammation has subsided. Correspondingly, the numbers of NPCs and OPCs in the DG and CC, respectively, of EAE-recovered mice increased compared to that of naive mice (NPCs, p 20 %) of the CXCL12(+) NPCs and OPCs co-express the CXCL12 receptor, CXCR4, and their numbers significantly increase with recovery from EAE not only relative to naive mice (p < 0.0002) but also to mice with ongoing EAE (p < 0.004). Conclusions: These data link CXCL12 expression in the DG and CC of EAE-recovering mice to the promotion of neuro/oligodendrogenesis generating CXCR4(+)CXCL12(+) neuronal and oligodendrocyte progenitor cells endowed with intrinsic neuro/oligondendroglial differentiation potential. These findings highlight the post-CNS-inflammation role of CXCL12 in augmenting the endogenous myelin/neuronal repair capacity in MS-like disease, likely via CXCL12/CXCR4 autocrine signaling

    Transcriptional response to interferon beta-1a treatment in patients with secondary progressive multiple sclerosis

    Get PDF
    Background: Interferon (IFN) beta-1a is an approved treatment for relapsing remitting multiple sclerosis (RRMS) and has been examined for use in secondary progressive multiple sclerosis (SPMS). However, no information regarding blood transcriptional changes induced by IFN treatment in SPMS patients is available. Our aim was to identify a subgroup of SPMS patients presenting a gene expression signature similar to that of RRMS patients who are clinical responders to IFN treatment. Methods: SPMS patients (n = 50, 20 IFN treated and 30 untreated) were classified using unsupervised hierarchical clustering according to IFN inducible gene expression profile identified in RRMS clinical responders to treatment. IFN inducible gene expression profile was determined by finding differentially expressed genes (DEGs) between IFN treated (n = 10) and untreated (n = 25) RRMS patients. Validation was performed on an additional independent group of 27 SPMS IFN treated patients by qRT-PCR. Results: One hundred and four DEGs, enriched by IFN signaling pathway (p = 7.4E-08), were identified in IFN treated RRMS patients. Classification of SPMS patients based on these DEGs yielded two patient groups: (1) IFN transcriptional responders (n = 12, 60 % of SPMS treated patients) showing gene-expression profile similar to IFN treated RRMS patients; (2) IFN transcriptional non-responders (n = 8) showing expression profile similar to untreated patients. IFN transcriptional responders were characterized by a more active disease, as defined by higher EDSS progression and annual relapse rate. Conclusion: Within the IFN treated SPMS population, 60 % of patients have a transcriptional response to IFN which is similar to that of RRMS patients who are IFN responders to treatment

    ‘Multi-Epitope-Targeted’ Immune-Specific Therapy for a Multiple Sclerosis-Like Disease via Engineered Multi-Epitope Protein Is Superior to Peptides

    Get PDF
    Antigen-induced peripheral tolerance is potentially one of the most efficient and specific therapeutic approaches for autoimmune diseases. Although highly effective in animal models, antigen-based strategies have not yet been translated into practicable human therapy, and several clinical trials using a single antigen or peptidic-epitope in multiple sclerosis (MS) yielded disappointing results. In these clinical trials, however, the apparent complexity and dynamics of the pathogenic autoimmunity associated with MS, which result from the multiplicity of potential target antigens and “epitope spread”, have not been sufficiently considered. Thus, targeting pathogenic T-cells reactive against a single antigen/epitope is unlikely to be sufficient; to be effective, immunospecific therapy to MS should logically neutralize concomitantly T-cells reactive against as many major target antigens/epitopes as possible. We investigated such “multi-epitope-targeting” approach in murine experimental autoimmune encephalomyelitis (EAE) associated with a single (“classical”) or multiple (“complex”) anti-myelin autoreactivities, using cocktail of different encephalitogenic peptides vis-a-vis artificial multi-epitope-protein (designated Y-MSPc) encompassing rationally selected MS-relevant epitopes of five major myelin antigens, as “multi-epitope-targeting” agents. Y-MSPc was superior to peptide(s) in concomitantly downregulating pathogenic T-cells reactive against multiple myelin antigens/epitopes, via inducing more effective, longer lasting peripheral regulatory mechanisms (cytokine shift, anergy, and Foxp3+ CTLA4+ regulatory T-cells). Y-MSPc was also consistently more effective than the disease-inducing single peptide or peptide cocktail, not only in suppressing the development of “classical” or “complex EAE” or ameliorating ongoing disease, but most importantly, in reversing chronic EAE. Overall, our data emphasize that a “multi-epitope-targeting” strategy is required for effective immune-specific therapy of organ-specific autoimmune diseases associated with complex and dynamic pathogenic autoimmunity, such as MS; our data further demonstrate that the “multi-epitope-targeting” approach to therapy is optimized through specifically designed multi-epitope-proteins, rather than myelin peptide cocktails, as “multi-epitope-targeting” agents. Such artificial multi-epitope proteins can be tailored to other organ-specific autoimmune diseases

    Additional file 2: Figure S2. of Post-CNS-inflammation expression of CXCL12 promotes the endogenous myelin/neuronal repair capacity following spontaneous recovery from multiple sclerosis-like disease

    No full text
    The CXCL12 expression, CXCL12+ NPCs and CXCL12+ OPCs in the DG and CC of mice that did not recover from EAE, compared to recovered mice or mice at the peak of the disease. Brain sections of mice that did not recover from EAE (non-recovered; clinical score 2) were taken at the end point of the experiment (day 65 post-immunization which is also close to the time points after immunization when EAE-recovered mice were taken for analysis; Fig. 1a). The sections were immmunostained for MAC2 (green) or CXCL12 (red), or for co-immunostained for CXCL12 and immune markers of astrocytes (GFAP), NPCs (DCX), or OPCs (NG2). The representative images from the immunostained sections from the non-recovered mice (n = 4) were placed for comparison side by side with the images from recovered mice, mice at the peak of the disease, and from naïve mice, which are presented in Fig. 2. Representative images show (a) immunostaining for MAC2 (green) in brain sections from the D3V. (b) Immunostaining for CXCL12 (red) in brain sections from CC (central/caudal regions, denoted by dashed lines). (c) Immunostaining for CXCL12 (red) in brain sections from the DG. (d) Co-immunostaining for GFAP (green) and CXCL12 (red) in brain sections from the DG. (e) Co-immunostaining for NPCs (DCX+, green) and CXCL12 (red) in the DG. (f) Co-immunostaining for OPCS (NG2+, green) and CXL12 (red) in CC. Nuclei were visualized by DAPI counterstaining (blue). The brain sections were from same immunizations. The images shown for the non-recovered mice were from immunostaining of preserved free-floating brain sections that were kept in the presence of NaN3 for 18 months. Scale bar: a–f, 50 μm. Arrowheads in f indicate OPCs (NG2+) co-expressing CXCL12. (TIF 9358 kb

    Additional file 1: Figure S1. of Post-CNS-inflammation expression of CXCL12 promotes the endogenous myelin/neuronal repair capacity following spontaneous recovery from multiple sclerosis-like disease

    No full text
    High expression of CXCL12 in the CNS of mice with chronic clinical EAE. (a, b) Immunofluorescent images of spinal cord tissue sections (L1–L6) from mice with ongoing EAE (day 50 after immunization; clinical score =3) and from naive mice (a) immunostained for CXCL12 (red) or (b) co-stained for CXCL12 (green) and GFAP (red). (c) Immunofluorescent image of the cortex of the mice in panel a immunostained for CXCL12 (red). In a and c, nuclei were counterstained with DAPI (blue). Scale bars: a and c, 100 μm; b, 20 μm. (TIF 8873 kb
    corecore